<em>Mir-497</em> Regulates Cisplatin Resistance of Human Gastric Cancer Cell Line by Targeting IGF1R, IRS1 and BCL2

Research Article

Austin J Med Oncol. 2015;2(2): 1017.

Mir-497 Regulates Cisplatin Resistance of Human Gastric Cancer Cell Line by Targeting IGF1R, IRS1 and BCL2

Mingfeng He¹, Jiaqi Qian², Qin Shi³, Jun Zhu4, Yin Ding5, Xin Zhou², Wei Zhu²*, Wenfang Cheng³ and Ping Liu²*

1Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China

2Department of Oncology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China

3Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China

4Department of Radiation Oncology, Jiangsu Cancer Hospital, No. 42 Bai Zi Ting, Nanjing 210009, PR China

5State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, No. 22 Hankou road, Nanjing 210093, PR China #Contributed Equally

*Corresponding author: Dr. Wei Zhu or Prof. Ping Liu

Received: November 15, 2014; Accepted: April 27, 2015; Published: April 29, 2015

Abstract

Purpose: Studies showed that drug resistance of gastric cancer cells could be modulated by the abnormal expression of miRNAs which targeted multiple cell signal pathways. Here we aimed to investigate the possible role of miR-497 in the development of cisplatin resistance in human gastric cancer cell line.

Methods: miRNA Quantitative real-time PCR was used to detect the different miRNAs expression level between drug resistant and parental cancer cells. MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was used to test the drug resistance phenotype changes of cancer cells via over or down regulation of miRNAs. Dual-luciferase activity assay was used to verify the target genes of miRNAs. Western blot analysis, Immunohistochemistry, Immunofluorescence staining, Cell proliferation assay, clonogenic assay and apoptosis assay were used to elucidate the mechanism of miRNAs on modulating drug resistance of cancer cells.

Results: miR-497 was significantly down-regulated in both gastric cancer tissues and various gastric cancer cell lines. Moreover, it was down-regulated in cisplatin-resistant gastric cancer cell line SGC7901/cisplatin (DDP) and the down-regulation of miR-497 was concurrent with the up-regulation of IGF1R/ IRS1 pathway related proteins, such as IGF1R, IRS1 and BCL2, compared with the parental SGC7901 cell line, respectively. In vitro drug sensitivity assay demonstrated that over-expression of miR-497 sensitized SGC7901/DDP cells to cisplatin. The luciferase activity of the above proteins 3’-untranslated regionbased reporters constructed respectively in SGC7901/DDP cells suggested that IGF1R, IRS1 and BCL2 were all the direct target genes of miR-497. Enforced miR-497 expression reduced its target proteins level, inhibited SGC7901/ DDP cells proliferation and sensitized SGC7901/DDP cells to DDP-induced apoptosis.

Conclusions: Our findings suggested that hsa-miR-497 could modulate cisplatin resistance of human gastric cancer cell line at least in part by targeting IGF1R/IRS1 pathway.

Keywords: miR-497; Cisplatin resistance; IGF1R/IRS1 pathway; Gastric cancer

Abbreviations

miRNAs: microRNAs; DDP: Cisplatin; MTT: (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide)

Introduction

Drug resistance is one of the leading cause of chemotherapy failure. Earlier studies have clarified several cytological mechanisms of drug resistance of cancer cells, such as increased efflux of hydrophobic drugs, decreased uptake of water-soluble drugs and various changes influencing the capacity of cytotoxic drugs to kill cells, including alterations in cell cycle and proliferation, enhanced DNA repair activity, defective apoptosis, altered metabolism of drugs, etc. [1-4]. Recently, people have realized that multiple paths could cause drug resistance phenotype of cancer cells, such as genetic changes including mutations, translocations, deletions and amplification of genes or promoter regions and epigenetic changes including aberrant DNA methylation, histone modifications and non-coding RNA expression, etc. [5,6]. Recent studies have also implicated that epigenetic mechanisms do not necessarily require a stable heritable genetic alteration, might play a more important role in acquired drug resistance of cancer cells, which is closely relevant to clinical practice [7].

The microRNAs (miRNAs) are a group of small non-coding RNAs, which are single-stranded and consist of 19–25 nucleotides (~22 nt). The basic mechanism of miRNA action is that miRNA can imperfectly bind to the 3’UTR of target mRNAs, resulting in translational repression or target mRNA cleavage [8]. Recent studies suggested that the acquisition of drug resistance by cancer cells might be regulated via the change in miRNAs levels [9-12]. Emerging evidence have shown that knock-down or re-expression of specific miRNAs by synthetic anti-sense oligonucleotides or miRNAs precusors or mimics could modulate drug resistance [12]. For instance, plently of miRNAs, such as miR-15b, miR-16 and miR- 181b, could sensitize drug-resistant gastric cancer cell lines to drug induced apoptosis at least in part by targeting BCL2 [13,14]. miR-21 was up-regulated in various chemoresistant cancer cells, including breast cancer, glioblastoma and gastric cancer, while transfection with a specific anti-miR-21 could sensitize cancer cells to anti-cancer drugs via regulating PDCD4, LRRFIP1 or PTEN [15-17].

In this study, we reported that miR-497 was significantly downregulated in both gastric cancer tissues and various gastric cancer cell lines. Moreover, it was down-regulated in cisplatin-resistant gastric cancer cell line SGC7901/cisplatin (DDP). We demonstrated that miR-497 might modulate cisplatin resistance of human gastric cancer cell line at least in part by targeting IGF1R/IRS1 pathway.

Material and Methods

Clinical samples

Endoscopic biopsy specimens including 24 cases of gastric cancer and 30 cases of non-tumor gastric mucosa collected from the First Affiliated Hospital of Nanjing Medical University between March 2012 and December 2012 were included. All the samples were confirmed by pathological examination and stored in liquid nitrogen for miRNA analysis. While another 20 tissue samples obtained from surgical specimens diagnosed with gastric cancer from May 2011 to December 2012 were used for Immunohistochemistry detection. Approval for this study was obtained from the medical ethics committee of the First Affiliated Hospital of Nanjing Medical University (reference number: 2011-SRFA-058).

Cell culture

Human gastric epithelium cell line GES-1 and gastric adenocarcinoma cell lines MKN45, MGC803, BGC823 and SGC7901 were purchased from the National Institute of Cells (Shanghai, China). Cisplatin-resistant variant SGC7901/DDP was obtained from KeyGEN Biotechnology Company (Nanjing, China). All the cells were cultured in RPMI-1640 medium supplemented with 10% fetal calf serum (Gibco BRL, Grand Island, NY) in a humidified atmosphere containing 5% CO2 at 37°C. To maintain the cisplatinresistant phenotype, cisplatin (DDP, with final concentration of 1 μg/ ml) was added to the culture media for SGC7901/DDP cells.

Quantitative real-time PCR analysis for miRNA

Both biopsy specimens and cells were isolated with Trizol reagent (Invitrogen, Carlsbad, CA) and miRNA fraction was further purified using a mirVana ™ miRNA isolation kit (Ambion, Austin, TX). The concentration and purity of the RNA samples were determined spectroscopically. Expression of mature miRNA was assayed using stem-loop RT followed by real-time PCR analysis [14]. The primers of reverse transcription and polymerase chain reaction were purchased from RiboBio Co., Ltd (Guangzhou, China) named Bulge-Loop ™ miRNA qRT-PCR Primer Set as previously described [18]. qRTPCR was performed according to the protocol of the primer set. PCR product amplification was detected by the level of fluorescence emitted by SYBR Green (SYBR® Premix Ex Taq™ II, TaKaRa) which intercalated into double stranded DNA [18]. U6 gene was used for normalizing the each sample. The ΔCt method was used for miRNA expression analysis of biopsy specimens. First, the cycle number at the threshold level of fluorescence (Ct) for each sample was determined. Next, the ΔCt value was calculated. The ΔCt value was the difference between the Ct value of miR-497 and the Ct value of U6: ΔCt = Ct (miR-497) - Ct (U6). The fold-change for miRNA from cells relative to each control cells was calculated using the 2-ΔΔCt method [14], where ΔΔCt =ΔCt MKN45, MGC803, BGC823, SGC7901 -ΔCt GES- 1 or ΔΔCt =ΔCt SGC7901/DDP –ΔCt SGC7901. PCR was performed in triplicate.

In vitro drug sensitivity assay

SGC7901/DDP and SGC7901 cells were plated in 6-well plates (6×105 cells /well), 100 nM of the miR-497 mimic or 100 nM miRNA mimic control were transfected in SGC7901/DDP cells, while 100 nM of the miR-497 inhibitor or 100 nM miRNA inhibitor control were transfected in SGC7901 cells, using lipofectamine 2000 (Invitrogen, Long Island, NY, USA) according to the manufacturer’s protocol, respectively. The miR-497 mimic, miRNA mimic control, 2’-O-methyl (2’-O-Me) modified miR-497 inhibitor and miRNA inhibitor control were chemically synthesized by Shanghai GenePharma Company (Shanghai, China). The sequence of each was shown in Supplementary data. 1. Twenty-four hours after transfection cells were seeded into 96-well plates (5×103cells/well) for next step experiment. After cellular adhesion, freshly prepared cisplatin (DDP) was added with the final concentration being 0.01, 0.1, 1 and 10 times of the human peak plasma concentration for cisplatin as previously described [14]. The peak serum concentrations of cisplatin was 2.0 μg/ml [14]. 48 hr after the addition of drugs, cell viability was assessed by MTT assay. The absorbance at 490 nm (A490) of each well was read on a spectrophotometer. The concentration at which cisplatin produced 50% inhibition of growth (IC50) was estimated by the relative survival curve. Three independent experiments were performed in quadruplicate.

Dual-luciferase activity assay

The 3’UTR of human IGF1R, IRS1 and BCL2 cDNA containing the putative target site for the miR-497 (sequence shown in Supplementary data. 2) was chemically synthesized and inserted at the XbaI site, immediately downstream of the luciferase gene in the pGL3-control vector (Promega, Madison, WI) by Integrated Biotech Solutions Co., Ltd (Shanghai, China), respectively. Twentyfour hours before transfection, cells were plated at 1.5×105 cells/ well in 24-well plates. 200 ng of pGL3-IGF1R-3’-UTR, pGL3-IRS1- 3’-UTR or pGL3-BCL2-3’-UTR plus 80 ng pRL-TK (Promega) were transfected in combination with 60 pmol of the miR-497 mimic or miRNA mimic control using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s protocol as described, respectively [14]. Luciferase activity was measured 24hr after transfection using the Dual Luciferase Reporter Assay System (Promega). Firefly luciferase activity was normalized to renilla luciferase activity for each transfected well. Three independent experiments were performed in triplicate.

Immunohistochemistry

Twenty tissue samples were obtained from surgical specimens diagnosed with gastric cancer, from May 2011 to December 2012 at the First Affiliated Hospital of Nanjing Medical University. Tissue samples were formalin-fixed and paraffin-embedded; 4-μm-thick sections were cut and stained by using the avidinbiotin complex method. After that, the slides were pretreated with microwaves for antigen retrieval in 10 mM citrate buffer (pH 6.0) and incubated in the primary antibody at 4°C overnight. The antibody of IGF1Ra (Catalog: sc-271606), IRS1 (Catalog: sc-559) and BCL2 (Catalog: BS1511) were purchased from Santa Cruz Biotechnology and Bioworld Technology, respectively. If the staining was uncertain, we repeated to confirm it. For the scoring of the above proteins, the slides were scored by two separate observers blinded to the clinical data. They evaluated the immunostainings of the slides under an optical microscope of a magnification of 400×. If there were any intra-observer differences, the slides were reevaluated to reach consensus. The staining intensity of the above proteins expression was scored on a scale of 1–3 as follows: 0 score for no staining; 1 for weak staining; 2 for moderate staining; and 3 for strong staining. The percentage of positive cancer cells was scored as follows: 0 score for 0 %; 0.1 for 1–9 %; 0.5 for 10–49 %; and 1.0 for 50 % or more. We multiplied the staining intensity by the proportion score of the percentage of positive cancer cells. Thus, we separated the patients into positive ones (the product >1) and negative ones (the product =1).

Immunofluorescence staining

GES-1, MKN45, MGC803, BGC823, SGC7901 and SGC7901/ DDP cells were grown on glass coverslips and fixed with 4% paraformaldehyde at 4°C for 15 min and were further permeabilised and blocked with 0.5% Triton X-100 and 5% bovine serum albumin in phosphate buffered saline (PBS) for 30 min. The coverslips were then exposed to primary antibodies as mentioned above at 40C overnight, followed by the appropriate secondary antibodies. The preparations were visualised using an Olympus IX70 fluorescence microscope.

Western blot analysis

SGC7901/DDP cells were plated in 6-well plates (6×105 cells / well), 72 hr after the transfection of miR-497 mimic or miRNA mimic control, cells were harvested and homogenized with lysis buffer. Total protein was separated by denaturing 10% SDS–polyacrylamide gel electrophoresis. Total protein of GES-1, MKN45, MGC803, BGC823, SGC7901 and SGC7901/DDP was also extracted and separated as described above. Western blot analysis was performed as described [14]. The primary antibodies for IGF1Ra (Catalog: sc-271606), IRS1 (Catalog: sc-559), IGF1R (Catalog: BS1183), BCL2 (Catalog: BS1511) and GAPDH (Catalog: BS6945) were purchased from Santa Cruz Biotechnology and Bioworld Technology, respectively. Protein levels were normalized to GAPDH. Fold changes were determined.

Cell proliferation assay

Six hours after the transfection of miR-497 mimic or miRNA mimic control, SGC7901/DDP cells were trypsinized and seeded into 96-well culture plates at a density of 5×103cells/well in growth medium supplemented with 10% serum. The MTT assay was performed 24, 48 and 72 hr post-transfection and absorbance was measured using a spectrophotometer at 490 nm. Each assay was performed in triplicate with three independent replicates.

Clonogenic assay

SGC7901/DDP cells were transfected with miR-497 mimic or miRNA mimic control as previously described and plated into 6-well plates at a density of 200 cells per well, incubated at 37°C for 2 weeks, fixed and stained with crystal violet. The mean ±SEM number of colonies containing > 50 cells were counted under a microscope from three independent replicates.

Apoptosis assay

SGC7901/DDP cells were plated in 6-well plates (6×105 cells / well). Twenty-four hours after the transfection of miR-497 mimic or miRNA mimic control as described above, cells were treated by DDP, with final concentration of 10 μg/ml, respectively. 48 hr after the treatment of DDP, flow cytometry was used to detect apoptosis of the transfected SGC7901/DDP cells by determining the relative amount of AnnexinV-FITC-positive- PI-negative cells as previously described [14], respectively.

Statistical Analysis

Each experiment was repeated at least 3 times. Numerical data were presented as mean±SD. The difference between means was analyzed with Student’s t test. All statistical analyses were performed using SPSS11.0 software (Chicago, IL).

Differences were considered significant when p < 0.01.

Results

miR-497 was significantly down-regulated in gastric cancer tissues and gastric cancer cell lines

We examined miR-497 expression in endoscopic biopsy specimens including 24 cases of gastric cancer and 30 cases of nontumor gastric mucosa using quantitative real-time PCR, to determine whether miR-497 expression was associated with gastric cancer. We found that miR-497 expression was significantly down-regulated in gastric cancer tissues, with an average 5.21-fold decrease, compared with the non-tumor gastric mucosa (Supplementary data. 3A). Meanwhile, we also found that miR-497 was significantly downregulated in human gastric adenocarcinoma cell lines MKN45, MGC803, BGC823 and SGC7901, compared with human gastric epithelium cell line GES-1, with the average 3.57-fold, 2.77-fold, 3.22- fold and 4.75-fold decrease, respectively (Supplementary data. 3B).

miR-497 was significantly down-regulated in cisplatinresistant gastric cancer cell line SGC7901/ DDP

Quantitative real-time PCR for miR-497 verified that miR-497 was also significantly down-regulated in cisplatin-resistant gastric cancer cell line SGC7901/DDP. The average decreased fold change was 1.85, compared with the parental SGC7901 cells (Figure 1).